You are on page 1of 10

AAPS PharmSciTech, Vol. 13, No.

4, December 2012 ( Ⓒ 2012)


DOI: 10.1208/s12249-012-9847-7

Research Article

Product Development Studies on Surface-Adsorbed Nanoemulsion


of Olmesartan Medoxomil as a Capsular Dosage Form

Sumita Singh,1 Kamla Pathak,1 and Vikas Bali1,2

Received 8 May 2012; accepted 22 August 2012; published online 11 September 2012
Abstract. The present study aimed at development of capsular dosage form of surface-adsorbed nano-
emulsion (NE) of olmesartan medoxomil (OLM) so as to overcome the limitations associated with
handling of liquid NEs without affecting their pharmaceutical efficacy. Selection of oil, surfactant, and
cosurfactant for construction of pseudoternary phase diagrams was made on the basis of solubility of drug
in these excipients. Rationally selected NE formulations were evaluated for percentage transmittance,
viscosity, refractive index, globule size, zeta potential, and polydispersity index (PDI). Formulation (F3)
comprising of Capmul MCM® (10% v/v), Tween 80® (11.25% v/v), polyethylene glycol 400 (3.75% v/v),
and double-distilled water (75% v/v) displayed highest percentage cumulative drug release (%CDR;
96.69± 1.841), least globule size (17.51±5.87 nm), low PDI (0.203±0.032), high zeta potential (−58.93±0.98
mV), and hence was selected as the optimized formulation. F3 was adsorbed over colloidal silicon dioxide (2
ml/400 mg) to produce free-flowing solid surface-adsorbed NE that presented a ready-to-fill capsule
composition. Conversion of NE to surface-adsorbed NE and its reconstitution to NE did not affect the in
vitro release profile of OLM as the similarity factor with respect to NE was found to be 66% and 73%
respectively. The
%CDR after 12 h for optimized NE, surface-adsorbed NE, and reconstituted NE was found to be 96.69±0.54,
96.07 ±1.76, and 94.78±1.57, respectively (p>0.05). The present study established capsulated surface-
adsorbed NE as a viable delivery system with the potential to overcome the handling limitations of NE.
KEY WORDS: bioavailability; nanoemulsion; olmesartan medoxomil; oral.

INTRODUCTION pump and thus augment the intestinal absorption of therapeu-


tic agents susceptible to P-gp-mediated efflux in the intestine
Olmesartan medoxomil (OLM) is an extensively used (10).
antihypertensive drug (1). By the action of aryl esterases, OLM has been formulated as a self-microemulsifying
situated in both intestine and plasma, it gets quickly de-esteri- drug delivery system (SMEDDS) using capryol 90, tween 20
fied upon oral administration in to an active metabolite, i.e., and tetraglycol (10:60:30, v/v/v) and Lee and his coworkers
olmesartan (2). Olmesartan is reported to be more helpful in reported an improved relative bioavailability of 170% com-
patients with essential hypertension in comparison to other pared to the suspension in male Sprague–Dawley rats (11).
angiotensin II receptor blockers with respect to decrease in SMEDDS as a delivery system have been reported to employ
ambulatory blood pressure (3). Commercially available tab- high concentration (30–60%) of surfactants that may lead to
lets of OLM exhibit reduced oral absorption leading to low cellular toxicity (12). Furthermore, precipitation of the hydro-
oral bioavailability of 25.6% (4). OLM is highly lipophilic phobic therapeutic agent formulated as SMEDDS and encap-
in nature (log p=4.31) which attributes to its low aqueous sulated in gelatin capsule may occur due to propensity of the
solu- bility. Poor aqueous solubility and efflux of hydrophobic volatile solvents used in formulation of SMEDDS to get trans-
ther- apeutic agents by means of drug resistance pump in ferred to the shell (13). One of the approaches to circumvent
the gastrointestinal tract contribute to their low these limitations is to deliver hydrophobic therapeutic agents
bioavailability (5,6). Oil to water nanoemulsion systems are in the form of a nanoemulsion formulation that like SMEDDS
reported to augment aqueous solubility of hydrophobic is also isotropic, thermodynamically stable, transparent (or
therapeutic agents by including them in the oil phase of the translucent) system of oil, water, and surfactants. Nanoemul-
nanoemulsion (7–9). Non-ionic surfactants such as Tween 80 sion (NE) possesses droplet size generally in the range of 10–
have been stated to be valuable pharmaceutical excipient in 100 nm but in contrast to SMEDDS, it employs a lesser
order to prevent the function of the drug-resistant P- amount (5–10%) of surfactant. Furthermore, NEs can be
glycoprotein (P-gp) efflux formulated with little energy input (heat or mixing), have a
long shelf life, and is characterized with simplicity of scale up
1
Department of Pharmaceutics, Rajiv Academy for Pharmacy, of the manufacturing process (14). Hence, it was hypothesized
Mathura, 281001Uttar Pradesh, India. that developing a nanoemulsion-based drug delivery system
2
To whom correspondence should be addressed. (e- mail: utilizing a non-ionic surfactant as the emulsifier would help to
vksbali@gmail.com)

1530-9932/12/0400-1212/0 Ⓒ 2012 American Association of Pharmaceutical Scientists 1212


Surface-Adsorbed NE of Olmesartan Medoxomil 1213

improve solubility and prevent efflux of OLM out of the


Construction of Pseudoternary Phase Diagram
intestine eventually leading to enhanced oral bioavailability.
Although NE is one of the finest modes of delivery for
On the basis of solubility study of drug, Capmul MCM
hydrophobic therapeutic agents, but due to liquid nature of the
was chosen as the oil phase, Tween 80 as the surfactant, and
dosage form, it would not be as accepted as the solid dosage
PEG 400 as the cosurfactant. Double-distilled water was used
form. Liquid dosage forms are normally associated with trans-
as the aqueous phase for the creation of pseudoternary
portation issues, instability problems, and poor palatability due
phase diagram. Surfactant and cosurfactant were mixed
to the lipid content (in case of NE). Moreover, as potent drugs
(Smix) in different volume ratios (1:1, 1:2, 1:3, 2:1, 3:1, 4:1).
are incorporated in NE formulation, dose variability due to
For each phase diagram, oil and specific Smix ratio was
handling problems in case of select patient population may
mixed in differ- ent volume ratios from 1:9 to 9:1 so that
lead to toxicity. To manage these challenges, many attempts
maximum ratios were enclosed for the study in order to
have been made to convert liquid formulations into the solid
define the boundaries of phases formed in the phase
dosage forms like capsule (15–17) and tablet (18).
diagram. Pseudoternary phase diagrams were developed by
Based on these considerations, the current study was
aqueous titration data using PCP Disso V2.08 software,
aimed at developing and characterizing NE of OLM so as to
Pune, India. Physical state of the blank NE was marked on a
improve dissolution rate-limited absorption of the drug
pseudo-three-component phase diagram in which one axis
using a low surfactant concentration and to convert the
signified the aqueous phase, the second signified oil, and
developed liquid NE into a solid unit dosage form by
the third signified Smix. A total of 14 formulations were
adsorbing it over an inert, solid adsorbent so as to develop a
selected from the NE regions of the phase diagram.
patient friendly dos- age form.

MATERIALS Evaluation of Blank NEs

OLM was obtained as a gift sample from Sun Pharma-


ceutical Industries (Sikkim, India). Capmul MCM® (glycerol Thermodynamic Stress Stability Studies
monodicaprylate) and Captex 100® were obtained as gift
samples from Abitech Corporation Limited (Janesvile, WI, Blank NE formulations were centrifuged at 3,500 rpm
USA). Labrafil M 1944 CS® (oleoyl macrogoglyceride), Lab- for 30 min and monitored for phase separation, creaming, or
rafil M 2125CS® (linoleoyl macrogolglycerides), Labrasol® crack- ing. Those formulations that did not show any phase
(caprylo caproyl macrogol-8-glyceride), Peceol® (glyceryl separation were subjected to heating–cooling cycle. Six cycles
ole- ate), Plurol oleique® (polyglycerol oleate), Lauroglucol between re- frigerator temperature (4°C) and 45°C with
90® (propylene glycol monolaurate), and Transcutol® P storage at each tem- perature for not less than 48 h were
(diethy- lene glycol monoethyl ether) were donated by performed. The formulations that passed heating–cooling
Gattefosse (Saint Priest, Cedex, France). Tween 80® cycles were subjected to three freeze–thaw cycles at
(polyoxyethylene sorbitan monooleate), Tween 20® temperature between −21°C and +25°C with storage at each
(polyoxyethylene sorbitan monolaurate), ethanol, and temperature for not less than 48 h. The formulations that
polyethylene glycol 400 (PEG 400) were obtained from S.D. passed the thermodynamic stress stability tests were further
Fine-Chemicals Ltd. (Mumbai, India). Colloidal silicon taken for dispersibility study in order to estimate the efficiency
dioxide and dialysis membrane (pore size of 25 Å) were of emulsification.
obtained from Hi Media Laboratories Pvt. Ltd (Mumbai,
India). All other chemicals and solvents were of analytical
reagent grade and were used without further purification. Dispersibility Test

The efficiency of emulsification of oral NE was assessed


METHODS using a standard USP XXII dissolution apparatus II. One
milliliter of each formulation was added to 500 ml of double-
Screening of Components distilled water at 37±0.5°C. A standard stainless steel dissolu-
tion paddle rotating at 50 rpm presented gentle agitation. The
Solubility of OLM in various vehicles (oils, surfactants, in vitro behavior of the formulation was visually assessed
and cosurfactants) was determined by shake flask method using the grading system. Grade A was given to the
whereby an excess amount of drug was added to 2 ml of the formulations which exhibited clear or bluish appearance within
selected vehicle and kept at 25±1°C in an isothermal bath 1 min, grade B included formulations with slightly less clear
shaker (Hicon, New Delhi, India) for 72 h to reach and bluish white appearance, grade C was given to the
equilibri- um. The samples were centrifuged (Remi Pvt. formulations with fine milky appearance within 2 min, grade D
Ltd., Vasai, India) at 3,000 rpm for 15 min. Supernatant was assigned to the formulations with dull, grayish white
was removed carefully and diluted suitably with ethanol having slightly oily appearance in more than 2 min, and graded
(95%, v/v). The samples were analyzed for drug content at E were the for- mulations with large oil globules present on the
257.8 nm using UV- visible spectrophotometer (Shimadzu, surface. For- mulations, from each Smix ratio investigated, were
Pharmaspec1700, Kyoto, Japan). The solubility of drug in selected on the basis of least Smix and suitable amount of oil
each component was calculated in triplicate and (0.5 ml) that could completely dissolve the required amount (5
mean±standard deviation (SD) was reported. mg) of drug. Care was taken to select the formulations which
passed the thermodynamic stress stability and dispersibility
test in grades A and B.
1214 Singh, Pathak and Bali

Preparation of Drug-Loaded NE
before study to ensure complete wetting of the membrane.
Two milliliters of optimized formulations was placed in pre-
Thermodynamically stable blank NEs were chosen for
treated dialysis bag and drug release was studied using USP
drug loading. Drug-loaded NEs were prepared by adding the
dissolution apparatus II (Hicon Enterprises, Delhi, India)
calculated amount of drug (10.02 mg/ml of OLM) to the oil
containing 500 ml of phosphate buffer (pH7.4) at 37±0.5°C.
phase and stirring in isothermal bath shaker until whole of the
The speed of the paddle was adjusted to 50 rpm. Two-milliliter
drug was dissolved. Then, Smix in a fixed proportion was
sample was withdrawn at regular time intervals (0, 0.5, 1, 2, 3,
added to fixed volume of oil containing drug to produce a
4, 5, 6, 7, 8, 9, 10, 11, and 12 h) and the volume withdrawn
clear mixture. This was followed by adding definite proportion
was replaced with the fresh medium. The release of drug from
of water (drop wise) and shaking slowly until a clear NE was
the NE formulations was compared against the pure drug
obtained.
suspen- sion. The samples were analyzed at 257.8 nm and the
percent- age cumulative drug release (%CDR) was calculated.
Characterization of the NE The analysis of the samples was done in triplicate.

Globule Size, Zeta Potential, and Polydispersibility Index


Transmission Electron Microscopy
Globule size of the NEs was determined by photon cor-
relation spectroscopy that analyzes the fluctuation in light Morphology of the oil droplets in the NE formulation was
scattering due to Brownian motion of the globules, using a visualized using CM 10 transmission electron microscope
Zetasizer ver. 6.01 (Malvern Instrument Ltd., UK). The for- (Mega View III FW, Philips, UK). NE formulation was diluted
mulation was subjected to 500 times dilution with double- 100 times and a drop was applied to 300 mesh copper grid.
distilled water and light scattering was monitored at 25°C at The grid was inverted and a drop of phosphotungstic acid
a 90° angle. Zeta potential was also measured using the (PTA) was applied to the grid for 10 s. Excess of PTA was
same instrument. The refractive index was kept at 1.33 and removed and grid was analyzed at 60–80 kV.
viscosity at 1.0 cps to mimic the values for pure water. Zeta
potential values were determined from the electrophoretic
Selection of Optimized NE
mobility of oil droplets.
Optimized formulation was selected on the basis of glob-
Percentage Transmittance, Refractive Index, and Viscosity ule size, zeta potential, polydispersity index (PDI), percent
drug content, and %CDR. The optimized NE was utilized
Percentage transmittance was determined spectrophoto- for the preparation of surface-adsorbed NE by adsorption
metrically. One milliliter of the formulation was diluted to 100 over a solid adsorbent.
times with double-distilled water and percentage transmit-
tance was measured against double-distilled water as blank
at 630 nm. Abbe’s type refractrometer (Jindal Instruments, Adsorption of NE Over a Solid Adsorbent
Ambala, India) was used to determine the refractive index.
Few drops of NE were smeared on the lower prism surface. The optimized NE, F3 (5 ml) was placed in a glass mortar
The eye piece cross-wire was adjusted so that a sharp demar- and colloidal silicon dioxide (400 mg) was added slowly and
cation line passes through the center having half light and half mixed gently to get the solid mass. The solid mass was passed
dark position. Then, readings were noted down from the scale. through the sieve (22 mesh size) to get uniform free-flowing
The viscosity of the formulations was determined without powder. The powder was stored over anhydrous calcium chlo-
dilution using Brookfield viscometer DV-II + Pro (Brookfield ride in a dessicator until further evaluation.
Engineering Laboratories, Inc, MA, USA) coupled with S-94
spindle at 100 rpm and 25±2°C.
Micromeritic and Rheological Characterization of Surface-
Drug Content Adsorbed NE

For the determination of drug content, 1 ml of the NE The flow properties of the surface-adsorbed NE were
determined by the following tests: (1) Carr’s compressibility
was taken in a 10 ml volumetric flask and shaken
vigorously with ethanol (95%, v/v) for 10 min. Finally, the index, (2) Hausner’s ratio, and (3) angle of repose. The sur-
face-adsorbed NE (5 g) was poured lightly into 50 ml measur-
volume was made up to 10 ml with the same solvent and
after proper dilution using ethanol, the drug content was ing cylinder. The powder was subjected to tapping until no
further change in volume was observed. Bulk density (Do) and
analyzed at
257.8 nm by UV spectrophotometer (Shimadzu, Pharmas- tapped density (Df) of the powder was calculated by dividing
the weight of the granules by its volume before and after
pec1700, Kyoto, Japan).
tapping, respectively. Percentage compressibility was comput-
ed by subtracting Do from Df and divided by Df and multiplied
In Vitro Drug Release
by 100, Hausner’s ratio was computed as Df divided by Do.
Angle of repose was measured by static funnel method. Angle
In vitro drug release was measured by dialysis bag
of the heap of granules (5 g) formed by passing through a
meth- od using a pretreated dialysis membrane (MWCO 12–
funnel placed at a height of 8 cm from the horizontal surface
14 kD). Himedia dialysis membrane (Himedia Laboratories
was measured using a protractor (17).
Pvt. Ltd., Mumbai, India) was kept in a normal saline
solution for 2 h
Surface-Adsorbed NE of Olmesartan Medoxomil 1215

Drug Content Determination


confirmed by its poor solubility of 0.0071 mg/ml in water
(polar solvent). However, the solubility of OLM was higher
Surface-adsorbed NE equivalent to 5 mg of OLM was
in nonpolar solvents. Consequently, the solubility of OLM was
dispersed in suitable quantity of ethanol (95%, v/v). The
found to be maximum (10.023 ± 1.517 mg/ml) in Capmul
sample was mixed thoroughly to ensure complete dissolution
MCM, a medium chain mono/diglyceride (Fig. 1). Higher
of drug in ethanol. The sample was centrifuged using centri-
solubility of drug in Capmul MCM may be attributed to the
fuge (Remi Pvt Ltd., Vasai, India) at 3,500 rpm for 15 min to
nonpolar nature of the poorly water-soluble drugs that sup-
separate colloidal silicon dioxide particles. The supernatant
ports their solubilization in oils like medium chain triglycer-
was suitably diluted and analyzed spectrophotometrically at
ides or mono- or diglycerides (7). High solubility of drug in oil
257.8 nm. Drug content was computed from the validated
is particularly advantageous in NE formulation. The higher
calibration curve of drug in ethanol (95%, v/v).
the solubility of the drug in oil phase, the lower will be the
volume of oil required to dissolve the single dose of drug. As a
Scanning Electron Microscopy result, less quantity of surfactant and cosurfactant may be
required for NE formulation. In addition to oil, the solubility
The morphological features of particles of colloidal sili- of drug in the surfactant is also important. The solubility of
con dioxide and surface-adsorbed NE were investigated by OLM was highest in Tween 80, among the examined surfac-
JEOL-5400 (Tokyo, Japan) scanning electron microscope. tants and PEG 400 among the examined cosurfactants. Thus,
Gold sputter coating of all the samples was done to render for the formulation of NE, Capmul MCM, Tween 80, and
the surface of particles electroconductive. The micrographs PEG 400 were selected as oil phase, surfactant, and cosurfac-
were viewed at ×100 and ×500 magnifications. tant, respectively.

Assessment of Reconstituted NE
Construction of Pseudoternary Phase Diagrams
The surface-adsorbed NE powder (420 mg) was resus-
The study of phase behavior helps to precisely character-
pended in 3.75 ml double-distilled water and shaken gently for
ize a phase boundary. Knowledge about the boundaries of the
5 min. The reconstituted NE (RF3) was characterized for
different phases as a function of composition variables can be
globule size and zeta potential using Zetasizer ver. 6.01 (Mal-
attained by preparing phase diagrams. Selection of oil, surfac-
vern Instrument Ltd., UK) as described earlier. The globule
tant, and the mixing ratio of oil to surfactant/cosurfactant
size was visualized by TEM as detailed previously.
mixture are vital for the NE formation (7). Low toxicity,
resistance to pH, and ionic strength changes are some of the
In Vitro Drug Release attributes that favor utilization of nonionic surfactants for the
NE formulations. It was observed that when Tween80 (surfac-
In vitro drug release study of surface-adsorbed NE was tant) was used alone, a significant zone of NE was obtained
performed by introducing the powder equivalent to 5 mg drug (Fig. 2a) but when PEG 400 (cosurfactant) was used along
into the release test media using USP dissolution apparatus with the surfactant in a ratio of 1:1 (Fig. 2b), a tremendous
II (Hicon Enterprises, Delhi, India) with paddle rotation decrease in the NE region was observed and 72.72% (v/v) of
of 50 rpm. The dissolution media consisted of 500 ml of oil could be emulsified using 16.66% (v/v) of Smix. However,
phos- phate buffer, pH7.4 maintained at 37±0.5°C and when the cosurfactant was utilized in 1:2 ratio (Fig. 2c), the
samples were withdrawn at predetermined time intervals. amount of oil that could be emulsified was 52.17% (v/v) using
The amount of drug release was estimated by measuring 34.78% (v/v) of Smix. On further increasing the proportion of
absorbance of the samples at 257.8 nm and the release cosurfactant in the Smix to 1:3 (Fig. 2d), a decrease in NE
profiles of surface- adsorbed NE and RF3 were evaluated region was observed with the maximum amount of oil that
for similarity against the release profile of F3 (optimized could be emulsified being reduced to 38.78% (v/v) using
NE). 52.17% (v/v) of Smix. With the Smix ratio of 1:4 (Fig. 2e),
further decrease in the NE region was observed and the
Statistical Analysis maximum amount of oil that could be emulsified was
36.36% (v/v).
The results were expressed as mean±SD and were ana- In addition to varying cosurfactant in the Smix, the effect
lyzed statistically by one-way analysis of variance (ANOVA) of varying the concentration of surfactant in the Smix from
using Graph Pad Prism V5.04 software (San Diego, CA, 1:1 to 2:1 (Fig. 2f) resulted in considerable increase in the
USA). NE region. On changing the concentration of surfactant in
Smix from 2:1 to 3:1 (Fig. 2g) and then to 4:1 (Fig. 2h), it
RESULTS AND DISCUSSION was observed that NE region did not change significantly. It
was also concluded that 63.63% (v/v) of oil could be
Screening of Components emulsified using 11.25% (v/v) of surfactant in comparison
to emulsifica- tion of 10% (v/v) of oil by using 60% (v/v) of
Solubility of the therapeutic agent in the oil phase of surfactant.
nanoemulsion governs the ability of the nanoemulsion formu-
lation to preserve the drug in solubilised form during its shelf Selection of NEs from Phase Diagram
life and after oral administration. Hence, the solubility of the
therapeutic agent in oils, surfactants, and cosurfactants was From the pseudoternary phase diagrams, 14 formulations
the most significant criterion for the screening of NE compo- were selected to fulfill the following criteria
nents. OLM is hydrophobic and less polar in nature that was
1216 Singh, Pathak and Bali

Fig. 1. Saturation solubility bar chart for selection of excipients for nanoemulsion of OLM
at 25±1°C

1. The proportion of oil used should be able to solubilize


formulations were subjected to different thermodynamic sta-
the drug (single dose) completely. One milligram of
bility tests such as centrifugation, heating–cooling cycles, and
OLM is dissolved easily in 0.1 ml of oil.
freeze–thaw cycles (Table I). The formulations that passed
2. The minimum concentration of the Smix used for that
these tests were selected for the dispersibility study to evalu-
amount of oil was taken.
ate the efficiency of emulsification.
3. The frequently used dose of the OLM is 5 mg (19).
Therefore, 5 mg was selected as the dose for the de-
Dispersibilty
velopment of NE formulation.
4. For convenience, 2 ml was selected as the volume of
the NE formulation, so that it can be increased or Gastrointestinal (GI) fluid is responsible for the
decreased as per the requirement. dilution of oral NE and results in the gradual desorption of
surfactant located at the oil–water interface. This process is
thermody- namically governed by the tendency of the
surfactant to pre- serve its concentration to its critical
Thermodynamic Stress Stability
micelle concentration. In order to evaluate the
emulsification efficiency of NE formula- tions upon infinite
Thermodynamically stability of nanoemulsion formula-
dilution in GI fluid, double-distilled water was used as a
tions differentiates them from emulsions that have kinetic dispersion medium. This selection was based on the report
stability which, NEs by virtue of their thermodynamic stability
corroborating insignificant difference in the behavior of NE,
do not exhibit phase separation, creaming, or cracking unlike prepared using nonionic surfactants, dispersed in either water
emulsions that finally phase separate (20). Thus, the selected
or simulated gastric or intestinal fluid (21,22). The formu-
lations that passed the dispersibility test in double-distilled
water

Fig. 2. Pseudoternary phase diagrams involving Capmul MCM, Tween 80, and PEG 400 as the oil, surfactant, and cosurfactant, respectively.
Ratio of surfactant to cosurfactant in a is 1:0, b 1:1, c 1:2, d 1:3, e 1:4, f 2:1, g 3:1, and h 4:1. Marked area oil in water NE region
Table I. Selection of Final Test Nanoemulsion Formulations by Thermodynamic Stress Stability Studies and Dispersibilty Test Su
rfa
Percentage v/v of NE components Observation ce
Recoded -
Formulation code Smix ratio Oil Smix Water Centrifugation Heating-cooling cycle Freeze-thaw cycle Dispersibilty Results formulations Ad
so
N1 1:0 10 40 50 ✓ ✓ ✓ Grade A Passed F1 rb
N2 1:0 10 38 52 ✓ ✓ Х Grade C Failed –
ed
N3 1:0 10 36 54 ✓ ✓ Х Grade C Failed –
N
N4 2:1 10 15 75 ✓ ✓ ✓ Grade A Passed F2
N5 2:1 10 42 48 Х – – Grade D Failed –
E
N6 2:1 10 40 50 Х – – Grade D Failed – of
N7 2:1 10 38 52 Х – – Grade D Failed – Ol
N8 2:1 10 37 53 Х – – Grade D Failed – m
N9 3:1 10 15 75 ✓ ✓ ✓ Grade A Passed F3 es
N10 3:1 10 43 47 ✓ Х – Grade C Failed – ar
N11 4:1 10 15 75 ✓ ✓ ✓ Grade B Passed F4 ta
N12 4:1 10 42 48 Х – – Grade D Failed – n
N13 4:1 10 40 50 Х – – Grade D Failed – M
N14 4:1 10 35 55 ✓ ✓ ✓ Grade B Passed F5 ed

✓ = Pass, Х = fail

Table II. Mean (±SD, n=3) Globule Size, Polydispersity Index (PDI), Zeta Potential, Drug Content, Refractive Index, Percentage Transmittance, and Viscosity of Final Test Formulations

Mean globule Mean zeta potential± Mean Drug content± Mean refractive Mean percentage Mean viscosity±
Formulation code size±SD (nm) Mean PDI±SD SD (mV) SD (%) index±SD transmittance±SD SD (cps)
F1 15.08±7.92 0.150±0.012 −33.51±0.21 85.66±1.42 1.332±0.004 88.67±0.13 291±0.78
F2 79.09±6.54 0.543±0.021 −48.72±0.92 89.40±2.04 1.389±0.002 92.75±0.64 286±0.34
F3 17.51±5.87 0.203±0.032 −58.93±0.98 95.60±1.23 1.334±0.003 99.78±0.23 254±0.35
F4 29.21±10.25 0.490±0.041 −58.32±1.2 91.77±3.21 1.374±0.001 98.74±0.08 293±0.21
F5 206.00±8.76 0.480±0.032 −56.51±1.09 86.24±2.72 1.362±0.013 96.88±0.05 319±1.04

12
17
1218 Singh, Pathak and Bali

regards to degree of repulsion between adjacent, similarly


charged globules in NE. Conventionally, zeta potential can
be positive or negative in the range of −30 to +30 mV. The
globules of OLM NEs displayed negative value of zeta poten-
tial (Table II) that could be due to the presence of negatively
charged free fatty acids component of oil used in the formu-
lation of nanoemulsion. Maximum value of zeta potential of
−58.93±0.98 mV was recorded for F3 closely followed by F4
and F5, and least zeta potential was documented for F1.
Similar pattern was also observed for drug content determi-
nation and hence, it can be inferred that the amount of drug
entrapped in the NE globule also influenced the magnitude of
charge on the globules. The polydispersity index of F1 formu-
lation was least (0.150±0.012) followed by F3 formulation as
0.203±0.032. Lower value of PDI is favorable, as it ensures
Fig. 3. In vitro drug release study of NE formulations in phosphate
buffer pH7.4 at 37±1±°C uniformity in size of nanoemulsion globules.

Percentage Transmittance, Refractive Index, and Viscosity


in grade(s) A and B were selected for further study as
these formulations were certain to remain as NE upon The value of percentage transmittance of formulations F1
dispersion in the aqueous environment of the GI tract to F4 was closer to 100% indicating that the formulations were
(Table I). Selected formulations were taken for globule clear and transparent (Table II). Among all the formulations,
size, zeta potential, PDI, refractive index, viscosity, per- F3 showed highest value of percentage transmittance (99.78±
centage transmittance, drug content, and in vitro drug 0.23%) which was significantly (p<0.001) higher in compari-
release determinations. son to other formulations. The refractive index that also meas-
ures transparency was 1.334±0.003 for F3 very close to that of
Characterization of NE water (1.334) and ensured its homogeneous character. The
viscosity analysis revealed low viscosity for all formulations
and F3 showed minimum viscosity of 254±0.35 cps. Thus, F3
Globule Size, Zeta Potential, and Polydispersibility Index
can be easily administered as a uniform dose.
Drug release and subsequent absorption are largely in-
fluenced by the globule size of NE (23). Rapid diffusion of In Vitro Drug Release
drug from smaller droplets into aqueous phase favors drug
dissolution. Globule size of the prepared NE was In vitro drug release profiles of OLM from NE formula-
determined and results are shown in Table II. As observed, tions (F1–F5) and pure drug suspension (S) are shown in
formulation F1 had the smallest globule size (15.08±7.92 nm) Fig. 3. The release of OLM from all the NE formulations
followed by F3 (17.51±5.87 nm). F1 formulation contained was higher than the release profile of S (47.38%±0.352 in
higher Smix ratio (40%) as compared to F3 (15%) that 12 h). Maximum %CDR of 96.69 ± 1.841 was displayed by
probably favored reduc- tion in globule size. Diameter of formulation F3 followed by F4 (90.53%±3.311) in 12 h.
the dispersed oil droplets of the NE was found to be much The reason for the differences in percentage cumulative
smaller than the diameter of smallest blood capillary (400 drug release of F3 and F4 can be correlated to the Smix
nm). This is advantageous as it avoids any probability of composition and its quantity in NE. The Smix in F3 was in
capillary blockage during transfer of the droplets in vivo. The the ratio of 3:1whereas in F4 it was 4:1, both present in
small size of the globules also favors long circulation time 15% by volume in respective NEs. As described in liter-
(12,24) ature, an increase in surfactant concentration results in
Charge on the oil droplets in NE is another attribute that decrease in the droplet size, but this phenomenon levels
should be investigated while studying the absorption of nano- off at a particular surfactant concentration whereby any
emulsion (25). Zeta potential also indicates NE stability with
Table III. Interpretation of Drug
Release Pattern of Nanoemulsion
Formulations

r2 Value

Formulation code Zero order First order Higuchi Pe


F1 0.9723 0.9217 0.9143 0.8
F2 0.9876 0.8791 0.9163 0.8
F3 0.9854 0.8764 0.9747 0.9
F4 0.9882 0.8794 0.9643 0.9
F5 0.9458 0.9173 0.8976 0.9
S 0.9256 0.8769 0.9043 0.8
a
Enhancement in %CDR after 12 h with respect to suspension
Surface-Adsorbed NE of Olmesartan Medoxomil 1219

Fig. 4. Scanning electron micrographs of a colloidal silicon dioxide and b surface-adsorbed NE

further increase in surfactant concentration results in a


issues, instability problems, and poor palatability due to
raise in droplet size (26). Stabilization of oil droplets by
the lipid content (in case of NE). Moreover, dose vari-
virtue of decrease in droplet size can be attributed to the
ability due to handling problems in case of select patient
localization of surfactant monolayer at the oil–water inter-
population may lead to toxicity. To manage these chal-
face (27). However, further rise in surfactant concentra-
lenges, liquid NE can be developed as a solid dosage form
tion leads to increased penetration of water into oil
by adsorbing on a highly porous solid with sufficient
droplets causing breakdown of oil droplets and resulting
adsorbing capacity and convert it to a free-flowing pow-
in bigger droplets (28). Consequently, F4 had a larger
der. Colloidal silicon dioxide, a highly porous solid with a
globule size of 29.21 ± 10.25 nm in comparison to 17.51
specific surface area of 200–400 m2/g was selected (29) for
±
the development of capsular dosage form of surface-
5.87 nm of F3 formulation that offered a larger surface
adsorbed NE.
area for the release of OLM. In F5, very high percentage
of Smix (35%, v/v) resulted in large globules (206 ±
8.76 nm) that offered low surface area and consequently Characterization of Surface-Adsorbed NE
low %CDR of 72.16% in 12 h. In F1 and F2, low Smix
ratios resulted in larger-sized globules and hence poor
release than F3. Modeling of the in vitro release data Micromeritic and Rheological Characterization
indicated zero order as the best-fit model (Table III).
On applying one-way ANOVA followed by Dunnett’s test, The bulk density of surface-adsorbed NE was found
a significant difference was observed in the release pro- to be 0.610 ± 0.103 g/cm3 and the tapped density was
files of F2, F4 (p <0.05), and F3 (p< 0.01) in comparison 0.7812 ± 0.052 g/cm3. Close values of bulk and tapped
to S (drug suspension). density will ensure uniform filling of capsule shell at
industrial scale. The Hausner’s ratio derived from bulk
and tapped density was found to be 1.287 ± 0.572. A
Selection and Development of Optimized Formulation
Hausner’s ratio of less than 1.25 indicates good flow
property of the granules (30) and slightly higher value of
Formulation F3 with highest %CDR of 96.69±1.841, least
Hausner’s ratio can be improvised by use of flow
globule size (17.51±5.87 nm), lower PDI value (0.203±0.032),
activators. The angle of repose that was 31.79°± 0.54,
and high zeta potential (−58.93±0.98 mV) was selected as the categorized the flow as passable (31) reconfirming the
optimized formulation and was considered as stable and results of Hausner’s ratio. Carr’s compressibility index
homogeneous formulation. Transmission electron micros- was found to be 28.073 ± 1.782% indicating compressible
copy of F3 revealed dark and spherical spots against a nature of the surface-adsorbed NE that presents an
light background and the droplet size revealed by TEM opportunity for tabletting of the surface-adsorbed NE.
was in conformity with the zeta sizing results. Although All these results proved that the surface-adsorbed NE
NE is one of the finest mode of delivery for hydrophobic powder can be aptly filled in the hard gelatin capsule as
therapeutic agent OLM, but due to liquid nature of the a solid unit dosage form.
dosage form, it is normally associated with transportation

Table IV. Comparative Characterization of Reconstituted Nanoemulsion (RF3) and Nanoemulsion


(F3)

Formulation code Mean globule size±SD (nm) Mean PDI±SD Mean zeta potential±SD (mV) Mean drug content±SD (%)
RF3 28.98±0.98 0.332±1.35 −35.90±0.45 92.77±2.16
F3 17.51±5.87 0.203±0.03 −58.93±0.98 95.60±1.23
1220 Singh, Pathak and Bali

Fig. 5. Transmission electron micrographs of a optimized nanoemulsion (F3) and b recon-


stituted nanoemulsion (RF3)

Scanning Electron Microscopy Dosage Form Development of Surface-Adsorbed NE and its


Evaluation
Colloidal silicon dioxide appeared to be spherical
particles approximating 100 μm (Fig. 4a). The micrograph On the basis of bulk density of surface-adsorbed NE,
of surface-adsorbed NE was not different from colloidal bulk volume 0.61 cm3 was calculated for surface-adsorbed
silicon dioxide indicating uniform adsorption of NE (F3) NE. This bulk volume can be suitably filled in size
over colloidal silicon dioxide (Fig. 4b). No unusual signs “0” capsule having body volume of 0.69 cm3 (32) for a
of precipitation/crystallization of drug/excipient on the sur- dose size of 5 mg of OLM. The in vitro drug release
face of adsorbent were recognizable. This confirms that profile (Fig. 6) of encapsulated surface-adsorbed NE
the NE was homogeneously adsorbed over colloidal sili- showed lower drug release in the initial first hour in
con dioxide. comparison to surface-adsorbed NE attributable to the
additional disintegration step of the capsule shell. Later
on, beyond 3 h, the release magnitude was similar to
Reconstitution of Surface-Adsorbed NE and Evaluation reconstituted NE (RF3) and NE(F3) till the 12th hour. The
similarity factor (f2) of the release profiles was calculated
The particles of surface-adsorbed NE are expected to using PCP Disso V2.08 software. The value of f2 between
be released from the capsule and on contact with body F3(reference) and surface-adsorbed NE (test) was found to
fluids, these should instantaneously re-form into NE quite be 66 and for F3 (reference) and reconstituted NE it was
similar to the initial formulation. To assess the found to be 73. A value of f2 factor between 50 and100
reconstitution abil- ity, specified quantity of surface- indicates similarity in given set of the reference and test
adsorbed NE was shaken with specified quantity of water to samples. Hence, the in vitro drug releases of F3, surface-
get RF3 that was char- acterized for globule size, adsorbed NE and RF3
polydispersity index, zeta poten- tial, and drug content.
Results of the characterization are given in Table IV.
Globule size, polydispersity index, zeta potential, and
percentage drug content of formulation RF3 were found to
be significantly different (p<0.05) in com- parison to
formulation F3. The morphology of the recon- stituted NE
is shown in Fig. 5b. As observed, the globules were
spherical but of slightly bigger in size than F3. The
colloidal silicon dioxide particles might have interfered
with the reconstitution of NE and consequently a higher
globule size, PDI were observed. Lowering of drug content
can be attributed to adsorption of OLM on colloidal silcon
diox- ide. The reconsitutional behavior of surface-adsorbed
NE is expected to be different in vivo. The gastrointestinal
fluid constituents containing biosurfactants in conjugation
with the peristaltic movements will probably facilitate the
recon- stitution of NE favoring lower globule size, probably
Fig. 6. In vitro drug release profiles of F3, surface-adsorbed NE, and
close to initial formulation F3. RF3 in phosphate buffer pH7.4 at 37±1±°C
Surface-Adsorbed NE of Olmesartan Medoxomil 1221

can be adjudged as similar. This study illustrates the 10. Shono Y, Nishihara H, Matsuda Y, Furukawa S, Okada N, Fujita
T, et al. Modulation of intestinal P-glycoprotein function by cre-
potential of encapsulated surface-adsorbed NE as a
mophor EL and other surfactants by an in vitro diffusion chamber
suitable solid unit dosage form to circumvent the handling method using the isolated rat intestinal membranes. J Pharm Sci.
limitations associated with SMEDDS or SNEDDS or NE of 2004;93:877–85.
water-insoluble therapeutic agents. 11. Lee BS, Kang MJ, Choi WS, Choi YB, Kim HS, Lee SK, et al.
Solubilized formulation of olmesartan medoxomil for enhancing
oral bioavailability. Arch Pharm Res. 2009;132:1629–35.
CONCLUSION 12. Bali V, Ali M, Ali J. Nanocarrier for the enhanced bioavailability
of a cardiovascular agent: in vitro, pharmacodynamic,
OLM nanoemulsion was successfully prepared by the pharmacokinet- ic and stability assessment. Int J Pharm.
aqueous titration method. Formulation containing capmul 2011;403:46–56.
13. Kohli K, Chopra S, Dhar D, Arora S, Khar RK. Self-emulsifying
MCM (10% v/v), tween 80 (11.25% v/v), PEG 400 (3.75% v/ drug delivery systems: an approach to enhance oral bioavailabil-
v), and double-distilled water (75% v/v) was optimized and ity. Drug Discov Today. 2010;15:958–65.
was converted to solid powder by adsorbing onto colloidal 14. Vior MCG, Monteagudo E, Dicelio LE, Awruch J. A compara-
silicon dioxide and encapsulated. The in vitro release profile of tive study of a novel lipophilic phthlocyanine incorporated into
NE formulation: photophysics, size, solubility and thermodynam-
the encapsulated system was statistically similar to ic stability. Dyes Pig. 2011;91:208–14.
nanoemulsion proving the performance efficacy of the 15. Mandawgade SD, Sharma S, Pathak S, Patrawale VB.
encapsulated system. However, the performance Development of SMEDDS using natural lipophile: application to
characteristics need in vivo evalua- tion. The developed system artemether deliv- ery. Int J Pharm. 2008;362:179–83.
can be considered as a novel solid unit dosage form of OLM 16. Date AA, Nagarsenker MS. Design and evaluation of self nano-
emulsifying drug delivery system (SNEDDS) for cefpodoxime
nanoemulsion that has the potential to circumvent the handling proxetil. Int J Pharm. 2007;329:166–72.
limitations associated with NE. 17. Patel AR, Vavia PR. Preparation and in vivo evaluation of
SMEDDS (self-microemulsifying drug delivery system) contain-
ACKNOWLEDGMENTS ing fenofibrate. AAPS J. 2007;9:E344–52.
18. Nazzal S, Khan MA. Controlled release of a self-emulsifying
formulation from a tablet dosage form: stability assessment and
The authors are thankful to Sun Pharmaceutical Indus- optimization of some processing parameters. Int J Pharm.
tries, Sikkim, India for providing the gift sample of olmesartan 2006;315:110–21.
medoxomil. 19. Sinko PJ. Martin’s pharmacy and pharmaceutical sciences. 5th ed.
Woltre Kulwer (India) Pvt Ltd.; 2008.
20. Laies P, Puchler K, Kirch W. The pharmacokinetics and metabol-
ic profile of olmesartan medoxomil limits the risk of clinically
REFERENCES relevant drug interaction. J Hypertens. 2001;19:S21–32.
21. Shinoda K, Kuneda H. The effect of salt concentration,
temperature, and additives on the solvent property of aerosol
1. Murakamia T, Konnoa H, Fukutsua N, Onoderaa M, Kawasakia T, OT solution. J Colloid Interface Sci. 1987;118:586–9.
Kusub F. Identification of a degradation product in stressed tablets 22. Shafiq S, Shakeel F, Talegaonkar S, Ahmad FJ, Khar RK,
of olmesartan medoxomil by the complementary use of HPLC Ali M. Development and bioavailability assessment of ram-
hyphenated techniques. J Pharmaceut Biomed Anal. ipril nanoemulsion formulation. Eur J Pharm Biopharm.
2008;47:553–9. 2007;66:227–43.
2. Park JH, Chang JS, El-Gamal MI, Choi WK, Lee WS, Chung HJ, 23. Khoo SM, Humberstone AJ, Porter CJH, Edwards GA, Charman
et al. Novel amides and esters prodrugs of olmesartan: synthesis, WN. Formulation design and bioavailability assessment of lipidic self-
bioconversion, and pharmacokinetic evaluation. Bioorg Med emulsi- fying formulations of halofantrine. Int J Pharm. 1998;167:155–
Chem Lett. 2010;20:5895–9. 64.
3. Klaus O, Stumpe MD. Olmesartan compared with other angioten- 24. Rajpoot P, Bali V, Pathak K. Anticancer efficacy, tissue distribu-
sin II receptor antagonists: head to head trial. Clin Ther. 2004;26: tion and blood pharmacokinetics of surface modified nanocarrier
A33–7. containing melphalan. Int J Pharm. 2012;426:219–30.
4. Wehling M. Can the pharmacokinetic characteristics of olmesartan 25. Constanitinides PP, Scalart JP, Lancaster C, Marcello J, Marks G,
medoxomil contribute to the improvement of blood pressure con- Ellens H. Formulation and intestinal absorption enhancement
trol? Clin Ther. 2004;26:A21–7. evaluation of water-in-oil microemulsions incorporating medi-
5. Nakagomi-Hagihara R, Nakai D, Kawai K, Yoshigae Y, Tokui um-chain glycerides. Pharm Res. 1994;11:1385–90.
T, Abe T. Oatp1b1, Oatp1b3 and Mrp2 are involved in 26. Gershanik T, Benzeno S, Benita S. Interaction of the self-
hepatobiliary transport of olmesartan, a novel angiotensin II emulsifying lipid drug delivery system with mucosa of
Blocker. Drug Metabol Dispos. 2006;34:862–9. everted rat intestine as a function of surface charge and
6. Matsushima S, Maeda K, Kondo C, Hirano M, Sasaki M, Suzuki droplet size. Pharm Res. 1998;15:863–9.
H, et al. Identification of the hepatic efflux transporters of organic 27. Yoo JH, Shanmugam S, Thapa P, Lee ES, Balakrishnan P, Yoo
anions using doubletransfected Madin-Darby canine kidney II SK. Novel self-nanoemulsifying drug delivery system for en-
cells expressing human organic anion-transporting polypeptide hanced solubility and dissolution of lutein. Arch Pharm Res.
1B1 (OATP1B1)/multidrug resistanceassociated protein 2, 2010;33:417–26.
OATP1B1/multidrug resistance 1, and OATP1B1/breast. J Phar- 28. Gursoy RN, Benita S. Self-emulsifying drug delivery system
macol Exp Ther. 2005;314:1059–67. (SEDDS) for improved oral delivery of lipophilic drugs. Biomed
7. Lawrence MJ, Rees GD. Microemulsion-based media as novel Pharmacother. 2004;58:173–82.
drug delivery system. Adv Drug Deliv Rev. 2000;45:89–121. 29. Pouton C. Formulation of self-emulsifying drug delivery systems.
8. Qian C, McClements DJ. Formation of nanoemulsions stabilized Adv Drug Deliv Rev. 1997;25:47–58.
by model food-grade emulsifiers using high-pressure homogeniza- 30. Row CR, Sheskey PJ, Owe SC. Handbook of pharmaceutical
tion: factors affecting particle size. Food Hydrocolloids. excipients. 5th ed. Royal society of Great Britain; 2006.
2011;25:1000–8. 31. Aulton ME. The science of dosage form design. 2nd ed. London:
9. Chen H, Khemtong C, Yang X, Chang X, Gao J. Nanonization Churchill & Livingstone; 2002.
strategies for poorly water soluble, drugs. Drug Discov Today. 32. Gennaro AR. Remington: the science and practice of pharmacy.
2011;16:354–60. 20th ed. Philadelphia: Lippincott Williams & Wilkins; 2004.

You might also like